• No results found

Trimethyllysine predicts all-cause and cardiovascular mortality in community-dwelling adults and patients with coronary heart disease

N/A
N/A
Protected

Academic year: 2022

Share "Trimethyllysine predicts all-cause and cardiovascular mortality in community-dwelling adults and patients with coronary heart disease"

Copied!
10
0
0

Laster.... (Se fulltekst nå)

Fulltekst

(1)

Trimethyllysine predicts all-cause and cardiovascular mortality in

community-dwelling adults and

patients with coronary heart disease

Espen Ø. Bjørnestad

1,

*, Indu Dhar

2

, Gard F.T. Svingen

3

, Eva R. Pedersen

3,4

,

Mads M. Svenningsson

3

, Grethe S. Tell

5

, Per M. Ueland

4

, Stein Ørn

1

, Gerhard Sulo

6

, Reijo Laaksonen

7

, and Ottar Nyga˚rd

2,3

1Department of Cardiology, Stavanger University Hospital, Gerd-Ragna Bloch Thorsens gate 8, 4011 Stavanger, Norway2Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Postboks 7804, 5020 Bergen, Norway3Department of Cardiology, Haukeland University Hospital, Jonas Lies vei 65, 5021 Bergen, Norway

4Department of Clinical Science, University of Bergen, Postboks 7804 NO-5020 Bergen, Norway5Department of Global Public Health and Primary Care, University of Bergen, A˚ rstadveien 17, 5020 Bergen, Norway6Centre for Disease Burden, Division of Mental and Physical Health, Norwegian Institute of Public Health, Zander Kaaesgate 7, 5015 Bergen, Norway7Finnish Cardiovascular Research Center, University of Tampere, Tampere University Hospital, Arvo Ylpo¨n Katu 34, 33520 Tampere, Finland

Received 27 April 2021; revised 14 June 2021; editorial decision 25 June 2021; accepted 28 June 2021 Handling Editor: Juan-Jesus Carrero

Aims Trimethyllysine (TML) is involved in carnitine synthesis, serves as a precursor of trimethylamineN-oxide (TMAO) and is associated with cardiovascular events in patients with established coronary heart disease (CHD). We pro- spectively examined circulating TML as a predictor of all-cause and cardiovascular mortality in community-dwelling adults and patients with CHD.

...

Methods and results

By Cox regression modelling, risk associations were examined in 6393 subjects in the community-based Hordaland Health Study (HUSK). A replication study was conducted among 4117 patients with suspected stable angina pectoris in the Western Norway Coronary Angiography Cohort (WECAC). During a mean follow-up of 10.5 years in the HUSK-cohort, 884 (13.8%) subjects died, of whom 287 from cardiovascular causes. After multivariable adjustments for traditional cardiovascular risk factors, the hazard ratio (HR) [95% confidence interval (95% CI)]

for all-cause mortality comparing the 4th vs. 1st TML-quartile was 1.66 (1.31–2.10,P< 0.001). Particularly strong associations were observed for cardiovascular mortality [HR (95% CI) 2.04 (1.32–3.15,P= 0.001)]. Corresponding risk-estimates in the WECAC (mean follow-up of 9.8 years) were 1.35 [1.10–1.66, P= 0.004] for all-cause and 1.45 [1.06–1.98, P= 0.02] for cardiovascular mortality. Significant correlations between plasma TML and TMAO were observed in both cohorts (rs>_ 0.42,P< 0.001); however, additional adjustments for TMAO did not materially influence the risk associations, and no effect modification by TMAO was found.

...

Conclusions Elevated TML-levels were associated with increased risk of all-cause and cardiovascular mortality both in subjects with and without established CHD.

䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏 䊏

* Corresponding author. Tel:þ4790037389, Email:espen.bjornestad@gmail.com

VCThe Author(s) 2021. Published by Oxford University Press on behalf of the European Society of Cardiology.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com

Downloaded from https://academic.oup.com/ehjopen/article/1/1/oeab007/6322956 by Universitetsbiblioteket i Bergen user on 27 January 2022

(2)

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.

Keywords Epidemiology

Risk factor

Cardiovascular outcomes

Mortality

Introduction

Growing evidence indicates that intestinal microbiota-derived metab- olites contribute to atherosclerosis, thrombosis, and systemic inflam- mation.1Particular interest has been given trimethylamineN-oxide (TMAO) as a potential mediator of cardiovascular disease (CVD).2 Trimethylamine N-oxide is generated in the liver from trimethyl- amine, which is produced by various gut microbes from dietary precursors such as phosphatidylcholine and L-carnitine.2 Dose- dependent associations of TMAO with cardiovascular outcomes have been observed in several high-risk patient cohorts;3,4however, the potential causative role of TMAO in atherothrombosis remains unclear.5

The TMAO-precursor trimethyllysine (TML) was recently identi- fied as a potential CVD risk marker in an untargeted metabolomics

approach.6Systemic TML is partly diet-derived from various plant- and animal sources.6,7Also, TML is produced endogenously during post-translational modifications of proteins, particularly histone methylation,8 which is a central epigenetic regulation process. In addition to its conversion to TMAO, TML is involved in several TMAO-independent pathways associated with CVD progression.9 Of note, observational clinical studies limited to patients with estab- lished CVD have found TML to be a predictor of future diabetes10 and cardiovascular events6,11,12beyond traditional risk factors.

The ability of TML to predict adverse outcomes in the general population is unknown. We therefore prospectively explored associ- ations between circulating TML and all-cause and cardiovascular mortality in a large Norwegian cohort of community-dwelling adults.

Further, we sought to validate the results in a higher-risk cohort of patients undergoing coronary angiography for stable angina pectoris.

Graphical Abstract

...

Downloaded from https://academic.oup.com/ehjopen/article/1/1/oeab007/6322956 by Universitetsbiblioteket i Bergen user on 27 January 2022

(3)

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

Methods .

Study design

Two independent cohorts of subjects with or without CHD were examined. The Hordaland Health study (HUSK) is a prospective, community-based cohort study of participants living in Western Norway and has been described in detail previously.13A total of 7051 subjects (born during 1925–27 or 1950–51) who underwent baseline surveys during 1997–99 were included. HUSK was the primary cohort in the current study.

The Western Norway Coronary Angiography Cohort (WECAC) is a prospective observational cohort study.14It includes 4164 patients who during 2000–04 underwent elective coronary angiography for suspected stable angina pectoris at two University Hospitals in Western Norway. Approximately two-third of the participants were enrolled in the Western Norway B-vitamin Intervention Trial (WENBIT; clinicaltrials.gov: NCT00354081), a randomized trial investigating effects of B-vitamin treatment on CVD outcomes and mortality.15

Subjects with missing data on plasma TML or covariables included in the risk models were excluded, yielding 6393 individuals in HUSK and 4117 patients in WECAC eligible for final analyses.

The study protocols for both HUSK and WECAC were approved by the Regional Committee for Medical and Health Research Ethics and the Norwegian Data Inspectorate. All study subjects provided written informed consent.

Baseline characteristics and biochemical analyses

The collection of baseline and biochemical data for both cohorts has been described in detail previously.13,14 Hypertension was defined by pre-existing diagnosis. Smoking was classified according to self-reported smoking habits and/or plasma cotinine concentrations >_ 85 nmol/L (avail- able in WECAC only). Diabetes mellitus included both types 1 and 2.

In HUSK, non-fasting venous blood samples were drawn at the base- line survey. In WECAC, blood samples were collected either 1–3 days before or immediately after the baseline angiography. Plasma TML and plasma TMAO were measured using liquid chromatography-tandem mass spectrometry (LC-MS/MS).16 Study-specific analyses for both HUSK and WECAC were performed at Bevital AS, Bergen, Norway (www.bevital.no), as previously described.17

Follow-up and clinical endpoints

For both cohorts, we obtained data on study endpoints from the Cause of Death Registry at Statistics Norway (http://www.ssb.no/en).18 A unique 11-digit personal identification number for each study subject was used to link to the registry. The primary outcomes were all-cause and cardiovascular mortality, whereas non-cardiovascular mortality was con- sidered as a secondary outcome. The study participants were followed from enrolment until death or throughout 2012.

Statistical analysis

Categorical variables are presented as percentages and continuous varia- bles as means [standard deviations (SD)]. Baseline characteristics of both cohorts were summarized across TML quartiles; trends were tested by linear regression for continuous variables, and logistic regression for or- dinal and binary variables. Baseline associations between TML and TMAO were additionally evaluated using Spearman rank correlations and scatter-plots. Survival was visualized using Kaplan–Meier curves, and dif- ferences across TML-quartiles were estimated with the log-rank test. In

addition, potential non-linear relationships between plasma TML, all-cause and cardiovascular mortality were visualized using a generalized additive model. Univariate, age- and sex-adjusted (Model 1) and multivari- able (Model 2) Cox regression models were applied to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). The multivariable model was adjusted for age (continuous), sex (binary), smoking (binary), dia- betes mellitus (binary), hypertension (binary), BMI (continuous), and total cholesterol (continuous). Additional adjustments for TMAO were per- formed in an extended model. Potential effect modifications by TMAO and risk factors in Model 2 were examined by adding interaction product terms to Model 1. Subgroups were created using existing categorical vari- ables or by separating continuous variables at the median level. As recent- ly recommended for observational studies, we applied E-value analyses to assess robustness to potential unmeasured or uncontrolled confounding.19

In the primary cohort (HUSK-cohort), we additionally assessed model fit when adding plasma TML to Model 2 by comparing the Akaikes’ infor- mation criterion.20Further, we tested model discrimination by calculating areas under receiver operator characteristics curves (ROC-AUC) and the integrated discrimination improvement (IDI). Reclassification of subjects was evaluated by calculating the continuous net reclassification improvement (NRI > 0).21

Results

Baseline characteristics (HUSK-cohort)

Baseline characteristics of the study participants according to plasma TML quartiles are presented inTable 1. HUSK consisted of 55.5%

women, mean (SD) age at baseline was 59 (12) years. Mean (SD) plasma TML and TMAO were 0.62 (0.30) and 9.11 (16.8)mmol/L, respectively.

There was a positive relationship between incremental TML quartiles and age, BMI, and creatinine. A higher proportion of subjects in the upper TML quartiles had hypertension and diabetes mellitus, whereas a lower proportion were current smokers.

No relationship was found with total cholesterol. Spearman’s correlation analysis showed a significant relationship between plasma TML and TMAO (rs = 0.44, P< 0.001) (Supplementary material online,Figure S1).

Plasma trimethyllysine and risk of mortality (HUSK-cohort)

A total of 884 (13.8%) subjects died over a mean (SD) follow-up of 10.5 (1.9) years, of whom 287 (32.4%) from cardiovascular and 597 (67.5%) from non-cardiovascular causes. Kaplan–Meier plots of event-free survival are presented inFigure 1. Using Cox regres- sion analyses, we found a graded positive association between plasma TML and mortality, particularly cardiovascular mortality (Table 2andFigure 2). The multivariable HRs (95% CI) comparing the 4th vs. 1st TML quartile were 1.66 (1.31–2.10,P< 0.001) and 2.04 (1.32–3.15,P= 0.001) for all-cause and cardiovascular mortal- ity, respectively. Notably, the risk relationships remained essential- ly unaltered after additional adjustments for TMAO [multivariable HRs (95% CI) of 1.72 (1.35–2.19P< 0.001) and 2.23 (1.43–3.48, P< 0.001) for all-cause and cardiovascular mortality, respectively].

Similarly, inclusion of creatinine in the multivariable model only slightly attenuated the risk estimates [HRs (95% CI) of

Downloaded from https://academic.oup.com/ehjopen/article/1/1/oeab007/6322956 by Universitetsbiblioteket i Bergen user on 27 January 2022

(4)

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. .

1.61 (1.25–2.07), P< 0.001] for all-cause and 1.88 (1.19–2.98, P= 0.01) for cardiovascular mortality.

Sensitivity analysis, model

discrimination, and reclassification (HUSK-cohort)

Application of E-value formula to Model 2 revealed E-values of 2.71 and 1.95 for the effect estimate and lower CI, when comparing the

4th vs. 1st plasma TML quartile in relation to all-cause mortality (Supplementary material online,Table S1). For CVD mortality, corre- sponding E-values were 3.50 for the effect estimate and 1.97 for the lower CI.

As shown in Supplementary material online, Table S2, the risk-relationship between plasma TML and all-cause mortality was not modified by conventional risk factors included in the multivariable model (pinteraction >_ 0.10), or by plasma TMAO (pinteraction= 0.90).

...

...

...

...

...

Table 1 Baseline characteristics of community-based subjects (HUSK-cohort) and patients with coronary heart dis- ease (WECAC) according to quartiles of plasma trimethyllysine

HUSK-cohort (n56393)

Quartile 1 (<0.46) Quartile 2 (0.46–0.55) Quartile 3 (0.55–0.68) Quartile 4 (>0.68) Ptrend

Plasma TML (mmol/L 0.41 (0.04) 0.50 (0.03) 0.61 (0.04) 0.94 (0.43) . . .

TMAO (mmol/L 4.53 (4.37) 5.78 (6.51) 7.93 (11.6) 18.2 (28.7) <0.001

Age (years) 54 (11) 58 (12) 60 (12.2) 62 (12.1) <0.001

Female sex (%) 85.0 62.4 41.8 33.9 <0.001

BMI (kg/m2) 24.8 (3.8) 25.5 (3.9) 26.0 (3.8) 26.5 (3.7) <0.001

Hypertension (%) 11.0 15.6 18.8 26.8 <0.001

Diabetes mellitus (%) 2.2 3.0 3.9 5.5 <0.001

Current smoking (%) 31.8 27.4 22.5 20.1 <0.001

Creatinine (mmol/L) 80.7 (8.6) 86.9 (9.6) 92.7 (11.0) 99.8 (17.3) <0.001

eGFR (mL/min per 1.73 m2) 75 (11) 72 (12) 70 (13) 66 (14) <0.001

Total cholesterol (mmol/L) 5.9 (1.1) 6.0 (1.1) 6.0 (1.1) 5.9 (1.1) 0.53

WECAC (n= 4117)

Quartile 1 (<0.54) Quartile 2 (0.54–0.67) Quartile 3 (0.67–87) Quartile 4 (>0.87) Ptrend

Plasma TML (mmol/L) 0.46 (0.06) 0.61 (0.04) 0.76 (0.06) 1.28 (0.70) . . .

TMAO (mmol/L) 5.15 (4.15) 6.92 (6.10) 8.28 (7.30) 15.75 (21.12) <0.001

Age (years) 60 (10) 61 (10) 62 (10) 64 (10) <0.001

Female sex (%) 47.1 30.6 18.7 16.3 <0.001

BMI (kg/m2) 25.9 (4.1) 26.2 (4.0) 26.3 (3.9) 26.8 (4.0) <0.001

Hypertension (%) 41.4 44.0 45.6 55.6 <0.001

Diabetes mellitus (%) 12.5 10.5 11.3 13.0 0.63

Current smoking (%) 34.8 32.7 29.2 30.2 0.01

Creatinine (mmol/L) 81.9 (10.9) 87.8 (11.8) 93.0 (12.7) 107.0 (51.7) <0.001

eGFR (mL/min per 1.73 m2) 95 (13) 91 (14) 87 (15) 79 (21) <0.001

Total cholesterol (mmol/L) 5.1 (1.1) 5.2 (1.3) 5.0 (1.1) 5.0 (1.2) 0.01

Prior CVD (%)

AMI 31.9 40.1 41.3 47.3 <0.001

PAD 6.4 8.1 9.9 11.6 <0.001

PCI 14.9 18.9 20.1 22.4 <0.001

CABG 8.7 10.2 11.5 15.6 <0.001

Medications after angiography (%)

Aspirin 79.4 81.0 82.1 83.4 0.02

Statins 77.4 79.5 82.3 81.3 0.01

CCB 20.5 20.4 21.6 27.5 <0.001

b-Blocker 69.3 71.4 73.9 74.8 0.002

ACEI 15.9 18.8 20.9 26.7 <0.001

Continuous variables are presented as means (SD), and categorical variables are reported as percentages.

ACEIs, angiotensin-converting-enzyme inhibitors; AMI, acute myocardial infarction; BMI, body mass index; CABG, coronary artery bypass grafting; CCB, calcium channel block- ers; CVD, cardiovascular disease; eGFR, estimated glomerular filtration rate; PAD, peripheral artery disease; PCI, percutaneous coronary intervention; TMAO, trimethylamine- N-oxide; TML, trimethyllysine.

Downloaded from https://academic.oup.com/ehjopen/article/1/1/oeab007/6322956 by Universitetsbiblioteket i Bergen user on 27 January 2022

(5)

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

For both total and cardiovascular mortality, the addition of plasma

.

TML to the multivariable model improved goodness of fit (Table 3).

As shown inTable 3, TML also improved model discrimination and reclassification of patients at risk.

Plasma trimethyllysine and risk

prediction among patients with coronary heart disease (WECAC)

Baseline characteristics of the WECAC are presented inTable 1.

Mean (SD) circulating TML was 0.77 (0.47) mmol/L. Similar to HUSK, incremental TML quartiles were positively associated with BMI and age, and patients in the upper TML-quartiles more likely had hypertension. In contrast to in the HUSK-cohort, no relationship was found with diabetes. TML and TMAO were

significantly correlated (rs = 0.42, P< 0.001) (Supplementary material online,Figure S2).

During a mean (SD) follow-up of 9.8 (2.6) years, 901 (21.9%) patients died, of whom 410 (45.5%) from cardiovascular causes and 491 (54.5%) from non-cardiovascular causes. Multivariable HRs (95% CI) comparing the 4th vs. 1st TML quartile were 1.35 (1.10–1.66, P= 0.004) and 1.45 (1.06–1.98,P= 0.02) for all-cause and cardiovascu- lar mortality, respectively (Table 4andFigure 1). The addition of TMAO to Model 2 slightly attenuated the risk estimates [multivariable HRs (95% CI) 1.29 (1.04–1.59), P= 0.02 and 1.37 (0.99–1.89), P= 0.06]

for all-cause and cardiovascular mortality, respectively]. Additional inclusion of creatinine attenuated the risk-associations to a greater extent [multivariable HRs (95% CI) 1.22 (0.99–1.50), P= 0.06] for all-cause and 1.30 (0.95–1.79,P= 0.10) for cardiovascular mortality].

No effect modification by TMAO was observed (pinteraction= 0.24).

(A)

1.00

(B)

(C) (D)

0.90

0.80

0.70

0 2 4 6

Time (Years)

Freedom from All-Cause Mortality

8 10 12

1.00

0.90

0.80

0.70

0.60

0 2 4 6

Time (Years)

8 10 12

1.00

0.95

0.90

0.85

0 2 4 6

Time (Years)

Freedom from CVD Mortality

8 10 12

1.00

0.95

0.90

0.85

0.80

0 2 4 6

Time (Years)

8 10 12

TML-Quartiles 1

Plog-rank < 0.001 2

3 4

TML-Quartiles 1

Plog-rank < 0.001 2

3 4

TML-Quartiles 1

Plog-rank < 0.001 2

3 4

TML-Quartiles 1

Plog-rank < 0.001 2

3 4

Quartile 1 1536 1526 1517 1502 1483 1447 1024 1007 992 962 932 623

Quartile 2 1648 1632 1611 1580 1537 1488 1017 995 970 928 878 597

Quartile 3 1614 1586 1552 1504 1450 1381 1067 1027 1001 962 916 580

Quartile 4 1595 1566 1519 1451 1397 1321 1009 960 922 870 812 513

Numbers at Risk

HUSK-Cohort WECAC

Figure 1Kaplan–Meier curves showing event-free survival according to quartiles of plasma trimethyllysine in the Hordaland Health Study-cohort (A and C) and Western Norway Coronary Angiography Cohort (B and D).

Downloaded from https://academic.oup.com/ehjopen/article/1/1/oeab007/6322956 by Universitetsbiblioteket i Bergen user on 27 January 2022

(6)

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. .

In a sensitivity analysis excluding patients (n= 1034) without any significant (>50%) stenotic coronary arteries at the baseline coronary angiography, we found essentially similar risk-associations [HRs (95%

CI) of 1.42 (1.14–1.78, P= 0.002) for total and 1.49 (1.07–2.06, P= 0.017) for cardiovascular mortality].

Discussion

In this large prospective two-cohort study including more than 10 000 subjects with or without established CHD, elevated circulating TML was reproducibly associated with a graded increased risk of long-term mortality, in particular cardiovascular mortality.

Trimethyllysine and cardiovascular disease

Existing data examining relationships of circulating TML with clinic- al outcomes are scarce and limited to high-risk patient populations.

We previously demonstrated plasma TML as a predictor of future myocardial infarction12in the WECAC. In a small study of patients with carotid atherosclerosis, TML was associated with an

increased 5-year risk of cardiovascular mortality.22 Similarly, positive associations of TML with cardiovascular events have recently been reported in patients with suspected chronic6and acute11coronary syndromes. The present study extends previous findings among patients with CVD and for the first time identifies TML as a predictor of all-cause and cardiovascular mortality in the general population. Thus, our results could suggest a role of TML in both primary and secondary risk prediction. However, a poten- tial clinical usefulness of the biomarker needs further exploration in future studies.

Trimethyllysine, trimethylamine N-oxide, and gut microbiota

Our observation of a strong association between plasma TML and TMAO in a community-based population validates findings from a recent report among patients presenting with the acute coronary syndrome.11Trimethyllysine contains a trimethylamine moiety and can be utilized as a substrate for microbiota-dependent TMAO-gen- eration.6However, its conversion to TMAO appears considerably lower compared to precursors such as choline and carnitine.6 ... ... ...

...

Table 2 Risk associations between plasma trimethyllysine and mortality in community-based subjects (HUSK-cohort)

Unadjusted Model 1a Model 2b

Events (%) HR (95%CI) P-value HR (95%CI) P-value HR (95%CI) P-value

All-cause mortality Plasma TML

Quartiles

Q1 6.9 Reference Reference Reference

Q2 11.2 1.67 (1.31–2.11) <0.001 1.17 (0.92–1.49) 0.19 1.20 (0.94–1.53) 0.14

Q3 16.9 2.59 (2.07–3.24) <0.001 1.47 (1.17–1.86) 0.001 1.59 (1.26–2.01) <0.001

Q4 20.1 3.15 (2.53–3.92) <0.001 1.51 (1.19–1.90) 0.001 1.66 (1.31–2.10) <0.001

Trend 1.44 (1.35–1.53) <0.001 1.14 (1.07–1.22) <0.001 1.18 (1.10–1.27) <0.001

Per 1-SDc 1.31 (1.24–1.38) <0.001 1.10 (1.03–1.18) 0.01 1.11 (1.04–1.19) 0.002

CVD mortality Quartiles

Q1 1.8 Reference Reference Reference

Q2 2.5 1.47 (0.91–2.36) 0.12 0.98 (0.61–1.59) 0.94 0.98 (0.61–1.58) 0.92

Q3 5.4 3.13 (2.05–4.79) <0.001 1.65 (1.06–2.56) 0.03 1.66 (1.07–2.58) 0.02

Q4 8.1 4.79 (3.18–7.21) <0.001 2.06 (1.34–3.19) 0.001 2.04 (1.32–3.15) 0.001

Trend 1.72 (1.53–1.93) <0.001 1.34 (1.18–1.52) <0.001 1.33 (1.17–1.51) <0.001

Per 1-SDc 1.44 (1.33–1.56) <0.001 1.24 (1.11–1.38) <0.001 1.19 (1.07–1.32) 0.002

Non-CVD mortality Quartiles

Q1 5.1 Reference Reference Reference

Q2 8.6 1.74 (1.32–2.30) <0.001 1.25 (0.95–1.66) 0.11 1.30 (0.98–1.72) 0.07

Q3 11.5 2.40 (1.84–3.12) <0.001 1.42 (1.07–1.86) 0.01 1.58 (1.19–2.08) 0.001

Q4 12.0 2.56 (1.97–3.33) <0.001 1.28 (0.97–1.70) 0.08 1.49 (1.12–1.98) 0.01

Trend 1.33 (1.23–1.43) <0.001 1.07 (0.98–1.16) 0.13 1.12 (1.03–1.22) 0.01

Per 1-SDc 1.24 (1.16–1.32) <0.001 1.03 (0.95–1.12) 0.47 1.07 (0.98–1.16) 0.12

CI, confidence interval; HR, hazard ratio; Q1, first quartile; Q4, fourth quartile; SD, standard deviation; TML, trimethyllysine.

aAdjusted for age and sex.

bAdjusted for age, sex, BMI, diabetes mellitus, current smoking, hypertension, and total cholesterol.

cLog-transformed.

Downloaded from https://academic.oup.com/ehjopen/article/1/1/oeab007/6322956 by Universitetsbiblioteket i Bergen user on 27 January 2022

(7)

.. ..

.. ..

.. ..

.. .

Notably, a microbiota-independent endogenous pathway generating TMAO from TML has also been suggested22 but currently lacks experimental evidence. In the present study, the risk estimates between circulating TML and mortality were only slightly attenuated after additional adjustments for TMAO, and there were no effect

modifications by TMAO. Thus, the TML-mortality relationship seems unlikely to be mediated solely through TMAO.

Intriguingly, a very recent animal study23unravelled an alternative gut-dependent metabolism of TML toN,N,N-trimethyl-5-aminovale- ric acid (TMAVA), which is implicated in the pathogenesis of the

(A) (B)

(C) (D)

Figure 2 Dose–response associations between plasma trimethyllysine and risk of total and cardiovascular mortality in the Hordaland Health Study-cohort (AandC) and Western Norway Coronary Angiography Cohort (BandD), adjusted for age and sex. The shaded areas covering the splines depicts 95% confidence intervals. Kernel density plots show the distribution of trimethyllysine.

...

Table 3 Model fit, discrimination, and reclassification in community-based subjects (HUSK-cohort)

AIC P-value NRI (95%CI) P-value IDI P-value ROC–AUC P-value

All-cause mortality

Modelawithout TML 4035 0.82

Modelawith TML 4016 <0.001 0.16 (0.09–0.23) <0.001 0.0046 <0.001 0.83 0.01

CVD mortality

Modelawithout TML 1894 0.84

Modelawith TML 1878 <0.001 0.35 (0.23–0.46) <0.001 0.0063 <0.001 0.85 0.03

AIC, Akaike’s information criterion; IDI, integrated discrimination index; NRI, net reclassification improvement.

aMultivariable model (Model 2) adjusted for age, sex, BMI, diabetes mellitus, current smoking, hypertension, and total cholesterol.

Downloaded from https://academic.oup.com/ehjopen/article/1/1/oeab007/6322956 by Universitetsbiblioteket i Bergen user on 27 January 2022

(8)

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

non-alcoholic fatty liver disease, a prevalent CVD risk factor.24 Accordingly, elevated TML-concentrations are present in patients with diagnosed liver steatosis and have been associated with increased risk of fatty liver.23Hence, several microbiota-dependent pathways may mediate the adverse relationship between TML and mortality.

Trimethyllysine, epigenetic regulation, and carnitine biosynthesis

Endogenous TML generation results from post-translational modifi- cations of proteins, such as histone methylation. This histone modifi- cation is dynamic and plays a potentially important regulatory role in pathways linked to CVD,25including endothelial dysfunction.26In this context, it is interesting that circulating TML predicted angiographic progression of atherosclerosis in a sub-cohort of WECAC.27 Of note, a strong correlation between free TML and protein-bound TML was reported in a small sample of both healthy subjects and sub- ject with CHD,6indicating that circulating free TML may partly reflect levels of protein-bound TML in the vasculature.

Trimethyllysine-availability regulates the rate of carnitine biosyn- thesis, with y-butyrobetaine as an intermediate.28Carnitines play an essential role for maintaining the mitochondrial function, and disrup- tions of carnitine homeostasis have been linked to decreased NO-sig- nalling and endothelial dysfunction.29 In line with data showing accumulation of carnitine pathway metabolites in obesity,10TML was positively associated with BMI in both cohorts. Notably, impaired car- nitine synthesis from TML has been suggested in the pathophysiology of both type 2-diabetes and atherothrombosis,10,12both major risk factors for mortality. However, the TML-carnitine-relationship is complex, as only about one-fourth of body carnitine sources are thought to come fromde novobiosynthesis.10Also, whether TML uti- lized for carnitine generation predominantly originates from en- dogenous or nutritional sources remains unclear.7,28

Trimethyllysine and renal function

Similar to TMAO, clearance of TML depends on renal excretion, and the kidney is an important regulator of carnitine biosynthesis.28 Additional adjustment for creatinine only mildly affected risk ... ... ...

...

Table 4 Risk associations between plasma trimethyllysine and mortality in patients with stable angina pectoris (WECAC)

Events (%) Unadjusted Model 1a Model 2b

HR (95%CI) P-value HR (95%CI) P-value HR (95%CI) P-value

All-cause mortality Plasma TML

Quartiles

Q1 15.1 Reference Reference Reference

Q2 20.4 1.36 (1.11–1.68) 0.004 1.10 (0.89–1.35) 0.39 1.10 (0.89–1.36) 0.38

Q3 22.7 1.56 (1.28–1.91) <0.001 1.13 (0.92–1.39) 0.25 1.14 (0.93–1.41) 0.21

Q4 29.4 2.12 (1.74–2.57) <0.001 1.38 (1.13–1.69) 0.002 1.35 (1.10–1.66) 0.004

Trend 1.27 (1.20–1.35) <0.001 1.11 (1.04–1.18) 0.001 1.10 (1.04–1.18) 0.002

Per 1-SDc 1.25 (1.18–1.33) <0.001 1.14 (1.07–1.22) <0.001 1.12 (1.05–1.19) 0.001

CVD mortality Quartiles

Q1 6.3 Reference Reference Reference

Q2 8.9 1.42 (1.04–1.96) 0.03 1.15 (0.83–1.58) 0.40 1.14 (0.82–1.57) 0.44

Q3 11.0 1.80 (1.33–2.43) <0.001 1.31 (0.96–1.79) 0.09 1.31 (0.96–1.79) 0.09

Q4 13.6 2.32 (1.72–3.11) <0.001 1.51 (1.11–2.05) 0.01 1.45 (1.06–1.98) 0.02

Trend 1.31 (1.20–1.43) <0.001 1.15 (1.04–1.26) 0.004 1.13 (1.03–1.24) 0.01

Per 1-SDc 1.28 (1.18–1.39) <0.001 1.17 (1.07–1.29) 0.001 1.14 (1.04–1.25) 0.01

Non-CVD mortality Quartiles

Q1 8.8 Reference Reference Reference

Q2 11.4 1.32 (1.00–1.74) 0.05 1.06 (0.80–1.40) 0.69 1.07 (0.81–1.42) 0.64

Q3 11.7 1.40 (1.06–1.83) 0.02 1.00 (0.76–1.32) 0.99 1.02 (0.77–1.35) 0.87

Q4 15.9 1.97 (1.53–2.56) <0.001 1.29 (0.98–1.68) 0.07 1.28 (0.98–1.68) 0.07

Trend 1.24 (1.14–1.34) <0.001 1.08 (0.99–1.18) 0.07 1.08 (0.99–1.18) 0.08

Per 1-SDc 1.23 (1.13–1.33) <0.001 1.11 (1.02–1.22) 0.02 1.10 (1.01–1.20) 0.03

CI, confidence interval; HR, hazard ratio; Q1, first quartile; Q4, fourth quartile; SD, standard deviation; TML, trimethyllysine.

aAdjusted for age and sex.

bAdjusted for age, sex, BMI, diabetes mellitus, current smoking, hypertension, and total cholesterol.

cLog-transformed.

Downloaded from https://academic.oup.com/ehjopen/article/1/1/oeab007/6322956 by Universitetsbiblioteket i Bergen user on 27 January 2022

(9)

.. ..

.. ..

.. ..

.. ..

estimates in the HUSK-cohort. Among patients in WECAC, how- ever, the risk estimates were attenuated to a greater extent numeric- ally, in particular for cardiovascular mortality. Renal disease is a major CVD risk factor;30hence, kidney function may limit the utility of TML as a prognostic marker in this patient population. As most subjects of both cohorts had adequate kidney function,31further studies among subjects with chronic kidney disease could further evaluate interac- tions of TML, renal function, and adverse outcomes.

Trimethyllysine and non-cardiovascular mortality

An interesting finding across both cohorts was the positive, albeit considerably weaker, relation of plasma TML with long-term non- cardiovascular mortality. Of note, proteins involved in both synthesis, removal and recognition of TML have been identified to contribute towards cancer development and several other diseases in humans.8 Also, the related metabolite TMAO has recently been linked to car- cinogenesis,32cognitive decline,33and adverse prognosis in patients with pulmonary disease.34Our findings need further replication and could motivate investigations on potential relationships of TML with non-cardiovascular disease outcomes, particularly in the general population.

Strengths and limitations

Major strengths of these prospective cohort studies include the large sample sizes, the replication of results in two independent popula- tions with different health profiles, the long-term follow-up and end- point data obtained from a national registry. We are aware of several limitations. First, both cohorts only included single measurements of plasma TML, which may have resulted in underestimation of risk associations due to regression dilution bias.35 Second, recognized challenges with using registry data to categorize causes of death in- clude differences in certification practices between physicians, use of unspecific medical codes, a low proportion of deaths in which medic- al autopsies are undertaken, and changes in coding practices over time.18However, a Norwegian autopsy study from 2011 showed a substantial agreement between mortality statistics and findings from autopsies for both coronary deaths and fatal strokes36Third, inher- ent to any observational study, the possibility of residual or unmeas- ured confounding cannot be ruled out. Of note, we found particularly high E-values for the association of TML with all-cause and cardiovascular mortality in the HUSK-cohort, indicating robust- ness to potential unmeasured confounding.19Fourth, both cohorts included mainly white subjects from a limited geographical area in Western Norway. As shown for TMAO, both geographical37and ethnic38 differences may considerably affect associations of microbiota-related metabolites with clinical outcomes. Our results, therefore, should be replicated in regions with different demograph- ics and nutritional profiles.

Conclusion

In two large prospective cohorts of subjects with or without established CHD, we observed consistent relationships of TML with long-term all-cause and cardiovascular mortality. Further research is

needed to clarify mechanisms determining systemic TML- concentrations.

Lead author biography

Espen Ø. Bjørnestad, MD, is a car- diology resident and researcher at the Department of Cardiology at Stavanger University Hospital. He has particular interest in bio- markers for cardiovascular risk pre- diction.

Supplementary material

Supplementary materialis available atEuropean Heart Journal Open online.

Acknowledgements

We thank the recruiting study personnel and other co-workers at Stavanger University Hospital, Stavanger, Norway and Haukeland University Hospital, Bergen, Norway. We are grateful to the staff of Statistics Norway and the Cause of Death Registry for providing outcome data on the study participants.

Funding

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Data availability

The data from WECAC that support the findings of this study are available from the corresponding author upon reasonable request.

Conflict of interest: none declared.

References

1. Witkowski M, Weeks TL, Hazen SL. Gut microbiota and cardiovascular disease.

Circ Res2020;127:553–570.

2. Tang WHW, Ba¨ckhed F, Landmesser U, Hazen SL. Intestinal microbiota in cardiovascular health and disease: JACC state-of-the-art review.J Am Coll Cardiol 2019;73:2089–2105.

3. Scvishhiattarella GG, Sannino A, Toscano E, Giugliano G, Gargiulo G, Franzone A, Trimarco B, Esposito G, Perrino C. Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: a systematic review and dose-response meta-analysis.Eur Heart J2017;38:2948–2956.

4. Yao M-E, Liao P-D, Zhao X-J, Wang L. Trimethylamine-N-oxide has prognostic value in coronary heart disease: a meta-analysis and dose-response analysis.BMC Cardiovasc Disord2020;20:7.

5. Zeisel SH, Warrier M. Trimethylamine N-oxide, the microbiome, and heart and kidney disease.Annu Rev Nutr2017;37:157–181.

6. Li XS, Wang Z, Cajka T, Buffa JA, Nemet I, Hurd AG, Gu X, Skye SM, Roberts AB, Wu Y, Li L, Shahen CJ, Wagner MA, Hartiala JA, Kerby RL, Romano KA, Han Y, Obeid S, Lu¨scher TF, Allayee H, Rey FE, DiDonato JA, Fiehn O, Tang WHW, Hazen SL. Untargeted metabolomics identifies trimethyllysine, a TMAO-producing nutrient precursor, as a predictor of incident cardiovascular disease risk.JCI Insight2018;3.

7. Servillo L, Giovane A, Cautela D, Castaldo D, Balestrieri ML. Where does N(e)-trimethyllysine for the carnitine biosynthesis in mammals come from?PLoS One2014;9:e84589.

Downloaded from https://academic.oup.com/ehjopen/article/1/1/oeab007/6322956 by Universitetsbiblioteket i Bergen user on 27 January 2022

(10)

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

.. ..

8. Maas MN, Hintzen JCJ, Porzberg MRB, Mecinovic J. Trimethyllysine: from

.

carnitine biosynthesis to epigenetics.Int J Mol Sci2020;21:9451.

9. Schiattarella GG, Trimarco B. Microbial metabolites as predictive biomarkers: a paradigm shift for cardiovascular risk stratification.Eur Heart J2019;40:2710–2712.

10. Strand E, Rebnord EW, Flygel MR, Lysne V, Svingen GFT, Tell GS, Løland KH, Berge RK, Svardal A, Nyga˚rd O, Pedersen ER. Serum carnitine metabolites and incident type 2 diabetes mellitus in patients with suspected stable angina pecto- ris.J Clin Endocrinol Metab2018;103:1033–1041.

11. Li XS, Obeid S, Wang Z, Hazen BJ, Li L, Wu Y, Hurd AG, Gu X, Pratt A, Levison BS, Chung Y-M, Nissen SE, Tang WHW, Mach F, Ra¨ber L, Nanchen D, Matter CM, Lu¨scher TF, Hazen SL. Trimethyllysine, a trimethylamine N-oxide precursor, provides near- and long-term prognostic value in patients presenting with acute coronary syndromes.Eur Heart J2019;40:2700–2709.

12. Bjørnestad E, Olset H, Dhar I, Løland K, Pedersen EKR, Svingen GFT, Svardal A, Berge RK, Ueland PM, Tell GS, Nilsen DWT, Nordrehaug JE, Nygaard E, Nyga˚rd O. Circulating trimethyllysine and risk of acute myocardial infarction in patients with suspected stable coronary heart disease.J Intern Med2020;288:446–456.

13. Refsum H, Nurk E, Smith AD, Ueland PM, Gjesdal CG, Bjelland I, Tverdal A, Tell GS, Nyga˚rd O, Vollset SE. The Hordaland Homocysteine Study: a community- based study of homocysteine, its determinants, and associations with disease.J Nutr2006;136:1731s–1740s.

14. Svingen GF, Ueland PM, Pedersen EK, Schartum-Hansen H, Seifert R, Ebbing M, Løland KH, Tell GS, Nyga˚rd O. Plasma dimethylglycine and risk of incident acute myocardial infarction in patients with stable angina pectoris.Arterioscler Thromb Vasc Biol2013;33:2041–2048.

15. Ebbing M, Bleie Ø, Ueland PM, Nordrehaug JE, Nilsen DW, Vollset SE, Refsum H, Pedersen EK, Nyga˚rd O. Mortality and cardiovascular events in patients treated with homocysteine-lowering B vitamins after coronary angiography: a randomized controlled trial.JAMA2008;300:795–804.

16. Midttun Ø, Kvalheim G, Ueland PM. High-throughput, low-volume, multianalyte quantification of plasma metabolites related to one-carbon metabolism using HPLC-MS/MS.Anal Bioanal Chem2013;405:2009–2017.

17. Bjørnestad E, Borsholm RA, Svingen GFT, Pedersen ER, Seifert R, Midttun Ø, Ueland PM, Tell GS, Bønaa KH, Nyga˚rd O. Neopterin as an effect modifier of the cardiovascular risk predicted by total homocysteine: a prospective 2-cohort study.J Am Heart Assoc2017;6:e006500.

18. Pedersen AG, Ellingsen CL. Data quality in the causes of death registry.Tidsskr nor Laegeforen2015;135:768–770.

19. VanderWeele TJ, Ding P. Sensitivity analysis in observational research: introduc- ing the E-value.Ann Intern Med2017;167:268–274.

20. Akaike H. A new look at the statistical model identification.IEEE Trans Automat Contr1974;19:716–723.

21. Pencina MJ, D’Agostino RB Sr, Steyerberg EW. Extensions of net reclassification improvement calculations to measure usefulness of new biomarkers.Stat Med 2011;30:11–21.

22. Skagen K, Trøseid M, Ueland T, Holm S, Abbas A, Gregersen I, Kummen M, Bjerkeli V, Reier-Nilsen F, Russell D, Svardal A, Karlsen TH, Aukrust P, Berge RK, Hov JE, Halvorsen B, Skjelland M. The carnitine-butyrobetaine-trimethylamine- N-oxide pathway and its association with cardiovascular mortality in patients with carotid atherosclerosis.Atherosclerosis2016;247:64–69.

23. Zhao M, Zhao L, Xiong X, He Y, Huang W, Liu Z, Ji L, Pan B, Guo X, Wang L, Cheng S, Xu M, Yang H, Yin Y, Garcia-Barrio MT, Chen YE, Meng X, Zheng L.

TMAVA, a metabolite of intestinal microbes, is increased in plasma from patients

with liver steatosis, inhibitsc-butyrobetaine hydroxylase, and exacerbates fatty liver in mice.Gastroenterology2020;158:2266–2281.e27.

24. Cai J, Zhang X-J, Ji Y-X, Zhang P, She Z-G, Li H. Nonalcoholic fatty liver disease pandemic fuels the upsurge in cardiovascular diseases. 2020;126:

679–704.

25. Udali S, Guarini P, Moruzzi S, Choi SW, Friso S. Cardiovascular epigenetics: from DNA methylation to microRNAs.Mol Aspects Med2013;34:883–901.

26. Kumar A, Kumar S, Vikram A, Hoffman TA, Naqvi A, Lewarchik CM, Kim YR, Irani K. Histone and DNA methylation-mediated epigenetic downregulation of endothelial Kruppel-like factor 2 by low-density lipoprotein cholesterol.

Arterioscler Thromb Vasc Biol2013;33:1936–1942.

27. Løland KH, Bleie O, Borgeraas H, Strand E, Ueland PM, Svardal A, Nordrehaug JE, Nyga˚rd O. The association between progression of atherosclerosis and the methylated amino acids asymmetric dimethylarginine and trimethyllysine.PLoS One2013;8:e64774.

28. Vaz FM, Wanders RJ. Carnitine biosynthesis in mammals.Biochem J2002;361:

417–429.

29. Sharma S, Black SM. Carnitine homeostasis, mitochondrial function, and cardio- vascular disease.Drug Discov Today Dis Mech2009;6:e31–e39.

30. Schiffrin EL, Lipman ML, Mann JF. Chronic kidney disease: effects on the cardio- vascular system.Circulation2007;116:85–97.

31. Zuo H, Svingen GFT, Tell GS, Ueland PM, Vollset SE, Pedersen ER, Ulvik A, Meyer K, Nordrehaug JE, Nilsen DWT, Bønaa KH, Nyga˚rd O. Plasma concentra- tions and dietary intakes of choline and betaine in association with atrial fibrilla- tion risk: results from 3 prospective cohorts with different health profiles.J Am Heart Assoc2018;7(8):e008190.

32. Chan CWH, Law BMH, Waye MMY, Chan JYW, So WKW, Chow KM.

Trimethylamine-N-oxide as one hypothetical link for the relationship between intestinal microbiota and cancer - where we are and where shall we go?J Cancer 2019;10:5874–5882.

33. Vogt NM, Romano KA, Darst BF, Engelman CD, Johnson SC, Carlsson CM, Asthana S, Blennow K, Zetterberg H, Bendlin BB, Rey FE. The gut microbiota- derived metabolite trimethylamine N-oxide is elevated in Alzheimer’s disease.

Alzheimer’s Res Ther2018;10:124.

34. Ottiger M, Nickler M, Steuer C, Bernasconi L, Huber A, Christ-Crain M, Henzen C, Hoess C, Thomann R, Zimmerli W, Mueller B, Schuetz P. Gut, microbiota- dependent trimethylamine-N-oxide is associated with long-term all-cause mor- tality in patients with exacerbated chronic obstructive pulmonary disease.

Nutrition2018;45:135–141.e1.

35. Clarke R, Shipley M, Lewington S, Youngman L, Collins R, Marmot M, Peto R.

Underestimation of risk associations due to regression dilution in long-term fol- low-up of prospective studies.Am J Epidemiol1999;150:341–353.

36. Gulsvik AK, Gulsvik A, Svendsen E, Mæhle BO, Thelle DS, Wyller TB. Diagnostic validity of fatal cerebral strokes and coronary deaths in mortality statistics: an autopsy study.Eur J Epidemiol2011;26:221–228.

37. Yazaki Y, Salzano A, Nelson CP, Voors AA, Anker SD, Cleland JG, Lang CC, Metra M, Samani NJ, Ng LL, Suzuki T. Geographical location affects the levels and association of trimethylamine N-oxide with heart failure mortality in BIOSTAT-CHF: a post-hoc analysis.Eur J Heart Fail2019;21:1291–1294.

38. Yazaki Y, Aizawa K, Israr MZ, Negishi K, Salzano A, Saitoh Y, Kimura N, Kono K, Heaney L, Cassambai S, Bernieh D, Lai F, Imai Y, Kario K, Nagai R, Ng LL, Suzuki T. Ethnic differences in association of outcomes with trimethylamine N-oxide in acute heart failure patients.ESC Heart Fail2020;7:2373–2378.

Downloaded from https://academic.oup.com/ehjopen/article/1/1/oeab007/6322956 by Universitetsbiblioteket i Bergen user on 27 January 2022

Referanser

RELATERTE DOKUMENTER

• The risk of all-cause and cardiovascular disease mortality was higher among people with more mental health problems, as scored on a mental health index, and a high alcohol intake

We aimed to evaluate 10 biomarkers related to inflammation and the kynurenine pathway, including neopterin, kynurenine:tryptophan ratio, C-reactive protein, tryptophan, and

OBJECTIVES The purpose of this study was to examine the joint and strati fi ed associations of sitting and MVPA with all-cause and cardiovascular disease (CVD) mortality, and to

In 1960, the Council of Europe took over responsibility for the work of the Universities Committee of the WEU and set up in its place a Com- mittee for Higher Education and

Unlike the Black Sea region, where Russia has recently used—and continues to use—military force and other means of influence in a concerted effort to redraw

The Baumol model predicts a steady increase in relative public sector prices (or costs) because of slow productivity growth and wage growth similar to sectors with higher

Next, we present cryptographic mechanisms that we have found to be typically implemented on common commercial unmanned aerial vehicles, and how they relate to the vulnerabilities

The Autodyn simulation code with the smooth particle hydrodynamic (SPH) method and Impetus Afea Solver with the corpuscular model are used and the results are compared with